Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Immunol ; 209(6): 1200-1211, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-35995508

RESUMEN

Dendritic cells (DCs) are professional APCs equipped with MHC-restricted Ags, costimulations, and cytokines that effectively prime and differentiate naive T cells into distinct functional subsets. The immune signals that DCs carry reflect the route of Ag uptake and the innate stimuli they received. In the mucosal tissues, owing to the great variety of foreign Ags and inflammatory cues, DCs are predominantly activated and migratory. In the small intestine, CD4 Th17 cells are abundant and have been shown to be regulated by DCs and macrophages. Using a mouse commensal bacteria experimental model, we identified that the early priming step of commensal-driven Th17 cells is controlled by bona fide Zbtb46-expressing DCs. CCR7-dependent migration of type 2 DCs (DC2s) from the small intestine to the mesenteric lymph nodes (MLNs) is essential for the activation of naive CD4 T cells. The migratory DC2 population in the MLNs is almost exclusively Esam+ cells. Single-cell RNA sequencing highlighted the abundance of costimulatory markers (CD40 and OX40) and chemokines (Ccl22 and Cxcl16) on MLN migratory DCs. Further resolution of MLN migratory DC2s revealed that the Th17-polarizing cytokine IL-6 colocalizes with DC2s expressing CD40, Ccl17, and Ccl22. Thus, early Th17 cell differentiation is initiated by a small subset of migratory DC2s in the gut-draining lymph nodes.


Asunto(s)
Células Dendríticas , Células Th17 , Bacterias , Quimiocinas , Citocinas , Interleucina-6 , Intestino Delgado , Ganglios Linfáticos , Membrana Mucosa , Receptores CCR7
2.
J Biol Chem ; 291(22): 11491-503, 2016 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-27008857

RESUMEN

Macrophages are pleiotropic cells capable of performing a broad spectrum of functions. Macrophage phenotypes are classified along a continuum between the extremes of proinflammatory M1 macrophages and anti-inflammatory M2 macrophages. The seemingly opposing functions of M1 and M2 macrophages must be tightly regulated for an effective and proper response to foreign molecules or damaged tissue. Excessive activation of either M1 or M2 macrophages contributes to the pathology of many diseases. Emodin is a Chinese herb-derived compound and has shown potential to inhibit inflammation in various settings. In this study, we tested the ability of emodin to modulate the macrophage response to both M1 and M2 stimuli. Primary mouse macrophages were stimulated with LPS/IFNγ or IL4 with or without emodin, and the effects of emodin on gene transcription, cell signaling pathways, and histone modifications were examined by a variety of approaches, including microarray, quantitative real-time PCR, Western blotting, chromatin immunoprecipitation, and functional assays. We found that emodin bidirectionally tunes the induction of LPS/IFNγ- and IL4-responsive genes through inhibiting NFκB/IRF5/STAT1 signaling and IRF4/STAT6 signaling, respectively. Thereby, emodin modulates macrophage phagocytosis, migration, and NO production. Furthermore, emodin inhibited the removal of H3K27 trimethylation (H3K27m3) marks and the addition of H3K27 acetylation (H3K27ac) marks on genes required for M1 or M2 polarization of macrophages. In conclusion, our data suggest that emodin is uniquely able to suppress the excessive response of macrophages to both M1 and M2 stimuli and therefore has the potential to restore macrophage homeostasis in various pathologies.


Asunto(s)
Polaridad Celular/efectos de los fármacos , Emodina/farmacología , Memoria Inmunológica/efectos de los fármacos , Inflamación/genética , Inflamación/inmunología , Macrófagos/inmunología , Animales , Células Cultivadas , Epigenómica , Humanos , Inflamación/tratamiento farmacológico , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/inmunología , Transducción de Señal/efectos de los fármacos
3.
J Lipid Res ; 57(6): 1006-16, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27063951

RESUMEN

Macrophage autophagy has been shown to be protective against atherosclerosis. We previously discovered that ursolic acid (UA) promoted cancer cell autophagy. In the present study, we aimed to examine whether UA enhances macrophage autophagy in the context of atherogenesis. Cell culture study showed that UA enhanced autophagy of macrophages by increasing the expression of Atg5 and Atg16l1, which led to altered macrophage function. UA reduced pro-interleukin (IL)-1ß protein levels and mature IL-1ß secretion in macrophages in response to lipopolysaccharide (LPS), without reducing IL-1ß mRNA expression. Confocal microscopy showed that in LPS-treated macrophages, UA increased LC3 protein levels and LC3 appeared to colocalize with IL-1ß. In cholesterol-loaded macrophages, UA increased cholesterol efflux to apoAI, although it did not alter mRNA or protein levels of ABCA1 and ABCG1. Electron microscopy showed that UA induced lipophagy in acetylated LDL-loaded macrophages, which may result in increased cholesterol ester hydrolysis in autophagolysosomes and presentation of free cholesterol to the cell membrane. In LDLR(-/-) mice fed a Western diet to induce atherogenesis, UA treatment significantly reduced atherosclerotic lesion size, accompanied by increased macrophage autophagy. In conclusion, the data suggest that UA promotes macrophage autophagy and, thereby, suppresses IL-1ß secretion, promotes cholesterol efflux, and attenuates atherosclerosis in mice.


Asunto(s)
Aterosclerosis/tratamiento farmacológico , Colesterol/metabolismo , Inflamación/tratamiento farmacológico , Interleucina-1beta/metabolismo , Triterpenos/administración & dosificación , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Animales , Aterosclerosis/metabolismo , Aterosclerosis/patología , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/genética , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/genética , Dieta Occidental , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/patología , Lipopolisacáridos/toxicidad , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Células RAW 264.7 , Receptores de LDL/genética , Ácido Ursólico
4.
Int J Cancer ; 136(6): E602-13, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25143000

RESUMEN

Immune cells in tumor microenvironment play a prominent role in tumor progression and metastasis. MicroRNA-155 (miR-155) represents an important player in innate and adaptive immunity by regulating differentiation, maturation and activation of macrophages, dendritic cells, B cells and T cells. However, the role of miR-155 expression in immune cells in solid tumor development is less elucidated. Our current study showed that both B16-F10 melanoma and Lewis lung carcinoma tumors grew much faster in bic/miR-155 knockout (miR-155(-/-) ) mice along with an increase of myeloid-derived suppressor cells (MDSCs) accumulation in tumors, compared to that in wild-type mice. Bone marrow transplantation study showed that bone marrow miR-155 deficiency could replicate the above tumor-promoting phenotype. In vitro study demonstrated that tumor-infiltrating miR-155(-/-) MDSCs showed greater migration ability and expressed higher level of multiple chemokines. Furthermore, we found that the level of HIF-1α, a direct target of miR-155, was increased in miR-155 deficient MDSCs, and that the increased HIF-1α upregulated CXCL1, CXCL3 and CXCL8 expression in MDSCs, contributing to the enhanced recruitment of miR-155(-/-) MDSCs to the tumors. Moreover, miR-155(-/-) MDSCs showed enhanced immunosuppressive and pro-angiogenic capacities. Taken together, our study, for the first time, demonstrated that miR-155 deficiency promoted solid tumor growth through increasing the recruitment of MDSCs to tumor microenvironment and enhancing the tumor-promoting functions of the recruited MDSCs. Thus, upregulating miR-155 expression in MDSCs may be developed as a therapeutic approach to halt tumor development.


Asunto(s)
MicroARNs/fisiología , Células Mieloides/fisiología , Neoplasias/patología , Microambiente Tumoral , Animales , Carcinoma Pulmonar de Lewis/patología , Movimiento Celular , Proliferación Celular , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL
5.
Breast Cancer Res Treat ; 148(2): 291-302, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25311112

RESUMEN

Breast cancer is the leading cause of death in female cancer patients due to the lack of effective treatment for metastasis. Macrophages are the most abundant immune cells in the primary and metastatic tumors, and contribute to tumor initiation, progression, and metastasis. Emodin has been found to exert anti-tumor effects through promoting cell cycle arrest and apoptosis, and inhibiting angiogenesis, but its effects on tumor-associated macrophages during cancer metastasis have not been investigated. Mice inoculated with 4T1 or EO771 breast cancer cells orthotopically were treated with Emodin after the primary tumors reached 200 mm3 in size. Primary tumor growth, lung metastasis, and macrophage infiltration in the lungs were analyzed. In vitro experiments were performed to examine the effects of Emodin on macrophage migration and M2 polarization, and the underlying mechanisms. Emodin significantly suppressed breast cancer lung metastasis in both orthotopic mouse models without apparent effects on primary tumors. Reduced infiltration of F4/80+ macrophages and Ym1+ M2 macrophages in lungs was observed in Emodin-treated mice. In vitro experiments demonstrated that Emodin decreased the migration of macrophages toward tumor cell-conditioned medium (TCM) and inhibited macrophage M2 polarization induced by TCM. Mechanistically, Emodin suppressed STAT6 phosphorylation and C/EBPß expression, two crucial signaling events in macrophage M2 polarization, in macrophages treated with IL-4 or TCM. Taken together, our study, for the first time, demonstrated that Emodin suppressed pulmonary metastasis of breast cancer probably through inhibiting macrophage recruitment and M2 polarization in the lungs by reducing STAT6 phosphorylation and C/EBPß expression.


Asunto(s)
Neoplasias de la Mama/prevención & control , Movimiento Celular/efectos de los fármacos , Emodina/uso terapéutico , Neoplasias Pulmonares/prevención & control , Macrófagos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Adhesión Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Front Oncol ; 11: 629846, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33898309

RESUMEN

During tumor development and progression, intrinsic and extrinsic factors trigger endoplasmic reticulum (ER) stress and the unfolded protein response, resulting in the increased expression of molecular chaperones to cope with the stress and maintain tumor cell survival. Heat shock protein (HSP) GRP94, also known as GP96, is an ER paralog of HSP90 and has been shown to promote survival signaling during tumor-induced stress and modulate the immune response through its multiple clients, including TLRs, integrins, LRP6, GARP, IGF, and HER2. Clinically, elevated expression of GRP94 correlates with an aggressive phenotype and poor clinical outcome in a variety of cancers. Thus, GRP94 is a potential molecular marker and therapeutic target in malignancies. In this review, we will undergo deep molecular profiling of GRP94 in tumor development and summarize the individual roles of GRP94 in common cancers, including breast cancer, colon cancer, lung cancer, liver cancer, multiple myeloma, and others. Finally, we will briefly review the therapeutic potential of selectively targeting GRP94 for the treatment of cancers.

7.
JCI Insight ; 6(17)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34283809

RESUMEN

Type 2 DCs (DC2s) comprise the majority of conventional DCs within most tumors; however, little is known about their ability to initiate and sustain antitumor immunity, as most studies have focused on antigen cross-presenting DC1s. Here, we report that DC2 infiltration identified by analysis of multiple human cancer data sets showed a significant correlation with survival across multiple human cancers, with the benefit being seen in tumors resistant to cytotoxic T cell control. Characterization of DC subtype infiltration into an immunotherapy-resistant model of breast cancer revealed that impairment of DC1s through 2 unique models resulted in enhanced DC2 functionality and improved tumor control. BATF3 deficiency depleted intratumoral DC1s, which led to increased DC2 lymph node migration and CD4+ T cell activation. Enhancing DC2 stimulatory potential by genetic deletion of Hsp90b1 (encoding molecular chaperon GP96) led to a similar enhancement of T cell immunity and improved survival in a spontaneous breast cancer model. These data highlight the therapeutic and prognostic potential of DC2s within checkpoint blockade-resistant tumors.


Asunto(s)
Células Dendríticas/inmunología , Regulación Neoplásica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/genética , Inmunidad Celular , Inmunoterapia/métodos , Melanoma Experimental/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Diferenciación Celular/inmunología , Línea Celular Tumoral , Reactividad Cruzada , Células Dendríticas/patología , Proteínas HSP90 de Choque Térmico/biosíntesis , Activación de Linfocitos , Melanoma Experimental/genética , Melanoma Experimental/terapia , Ratones Transgénicos , Neoplasias Experimentales , ARN Neoplásico/genética , Linfocitos T Citotóxicos/patología
8.
Redox Biol ; 37: 101609, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32591281

RESUMEN

Tristetraprolin (TTP), an mRNA binding and decaying protein, plays a significant role in controlling inflammation by decaying mRNAs encoding inflammatory cytokines such as TNFalpha. We aimed to test a hypothesis that TTP in bone marrow (BM) cells regulates atherogenesis by modulating inflammation and lipid metabolism through the modulation of oxidative stress pathways by TTP target genes. In a BM transplantation study, lethally irradiated atherogenic LDLR-/- mice were reconstituted with BM cells from either wild type (TTP+/+) or TTP knockout (TTP-/-) mice, and fed a Western diet for 12 weeks. We made the following observations: (1) TTP-/- BM recipients display a significantly higher systemic and multi-organ inflammation than TTP+/+ BM recipients; (2) BM TTP deficiency modulates hepatic expression of genes, detected by microarray, involved in lipid metabolism, inflammatory responses, and oxidative stress; (3) TTP-/- BM derived macrophages increase production of mitochondrial reactive oxygen species (mtROS); (4) BM-TTP-/- mice display a significant reduction in serum VLDL/LDL levels, and attenuated hepatic steatosis compared to controls; and (5) Reduction of serum VLDL/LDL levels offsets the increased inflammation, resulting in no changes in atherosclerosis. These findings provide a novel mechanistic insight into the roles of TTP-mediated mRNA decay in bone marrow-derived cells in regulating systemic inflammation, oxidative stress, and liver VLDL/LDL biogenesis.


Asunto(s)
Especies Reactivas de Oxígeno , Receptores de LDL , Tristetraprolina , Proteínas Adaptadoras Transductoras de Señales , Animales , Médula Ósea/metabolismo , Femenino , Humanos , Inflamación/genética , Lipoproteínas , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estabilidad del ARN , ARN Mensajero/genética , Receptores de LDL/genética , Tristetraprolina/genética , Tristetraprolina/metabolismo
9.
Int Immunopharmacol ; 75: 105763, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31325728

RESUMEN

Emodin is a Chinese herb-derived compound that exhibits a variety of pharmacological benefits. Although emodin has been shown to inhibit growth of cancer cells, its antineoplastic function is incompletely understood. CD155 is a member of poliovirus receptor-related (PRR) family of adhesion molecules; it is constitutively expressed on many tumor cell lines and tissues and has diverse functions. CD155 has been reported to mediate activation of T cells via CD226 or inhibition of T cells via T-cell immunoreceptor with Ig and ITIM domains (TIGIT). In addition, CD155 may play a critical role through non-immunological mechanisms in cancer. In this study, we tested the ability of emodin to modulate CD155 expression in cancer cells. We found that emodin significantly decreased the expression of CD155 in tumor cells and inhibited tumor cell proliferation and migration, and induced cell-cycle arrest at G2/M phase. The tumor inhibitory effects of emodin were lost with CD155 knockdown. Furthermore, emodin was used to treat mice bearing B16 melanoma. It was shown that emodin attenuated tumor growth accompanied by suppressing CD155 expression. Therefore, we propose that emodin could inhibit tumor growth, and the antineoplastic properties of emodin are at least partially CD155 dependent. Our study provides new insights into the mechanisms by which emodin inhibits tumor growth.


Asunto(s)
Antineoplásicos/farmacología , Emodina/farmacología , Melanoma Experimental/genética , Receptores Virales/genética , Neoplasias Cutáneas/genética , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Emodina/uso terapéutico , Femenino , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Ratones Endogámicos C57BL , ARN Interferente Pequeño/genética , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Carga Tumoral/efectos de los fármacos
10.
Innate Immun ; 25(4): 235-243, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31018807

RESUMEN

Dendritic cells (DCs) are professional Ag-presenting cells that play a critical role in both innate and adaptive immune responses. DCs recognize and respond to bacteria through multiple PRRs, including TLRs. Heat shock protein gp96/grp94 is a master essential chaperone for TLRs in the endoplasmic reticulum. We generated DC-specific gp96-knockout (KO) mice and showed that gp96 KO DCs were unable to respond to multiple TLR ligands. TLR-mediated hyperinflammatory response can lead to sepsis. However, the roles of neither DCs nor the DC-intrinsic gp96 in the process are completely understood. In a LPS-induced sepsis model, we hereby found that deletion of gp96 in DCs significantly reduced serum TNF-α levels and improved survival. Furthermore, using the well-defined polymicrobial sepsis model of cecal ligation and puncture, we found that DC-specific ablation of gp96 improved survival with significantly attenuated liver and renal injuries, decreased circulating inflammatory cytokines, altered DC maturation and activation, and increased serum Ig. Collectively, we demonstrate that deletion of gp96 in DCs is beneficial in protecting mice against sepsis induced by both endotoxemia and polymicrobial infections. We conclude that targeting gp96 in DCs may provide a potential novel approach for reducing the morbidity and mortality of sepsis.


Asunto(s)
Células Dendríticas/inmunología , Proteínas HSP70 de Choque Térmico/metabolismo , Riñón/patología , Hígado/patología , Proteínas de la Membrana/metabolismo , Sepsis/inmunología , Animales , Presentación de Antígeno , Ciego/cirugía , Diferenciación Celular , Proteínas HSP70 de Choque Térmico/genética , Humanos , Inmunidad Humoral , Mediadores de Inflamación/sangre , Lipopolisacáridos , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Modelos Animales , Factor de Necrosis Tumoral alfa/metabolismo
11.
Sci Rep ; 7(1): 2171, 2017 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-28526855

RESUMEN

The intestinal immunity and tolerance are orchestrated by both the innate and the adaptive immune system. Intestinal professional antigen presenting cells (pAPCs) recognize and respond to the gut microbiota through multiple pattern-recognition receptors, including TLRs and NLRs. How gut pAPCs maintain mucosal homeostasis remains incompletely understood. Heat shock protein gp96, also known as grp94, is an essential immune chaperone for TLRs. However, the role of gp96 in regulating CD11c+ APCs in the gut immunity and tolerance is unknown. By a genetic strategy, we report here that selective deletion of gp96 from CD11c+ cells in mice results in alteration of dendritic cell and T cell subsets in the gut as well as loss of antigen-specific regulatory T cell induction in the mesenteric lymph nodes. Strikingly, these conditional gp96-null mice developed spontaneous colitis, had increased levels of systemic and fecal IgA, and were highly susceptible to chemical-induced colitis. Our findings for the first time demonstrate that gp96 is essential for CD11c+ cells to induce regulatory T cells and maintain gut homeostasis, illustrating the importance of protein immune chaperone in safeguarding against immune pathology.


Asunto(s)
Antígeno CD11c/metabolismo , Tracto Gastrointestinal/metabolismo , Homeostasis , Glicoproteínas de Membrana/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Antígeno CD11c/genética , Colitis/etiología , Colitis/metabolismo , Colitis/patología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Epítopos de Linfocito T/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/patología , Eliminación de Gen , Inmunoglobulina A/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología
12.
Oncoimmunology ; 6(5): e1312042, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28638736

RESUMEN

Tumor microenvironment (TME) contains a variety of infiltrating immune cells. Among them, tumor-associated macrophages (TAMs) and their alternative activation contribute greatly to the progression of tumors. The mechanisms governing macrophage polarization in the TME are unclear. Here, we show that in TAMs or macrophages under tumor-conditioned medium treatment, the expression of transcription factor EB (TFEB) is reduced and more of the TFEB protein is in an inactive cytosolic form. Transforming growth factor (TGF)-ß is identified as a main driving force for the reduced TFEB expression and activity in TAMs via activating ERK signaling. TFEB interference in macrophages significantly enhanced their alternative activation, with reduced expression of MHC-II and co-stimulatory molecule CD80, decreased ability to activate T cells, and increased ability to attract tumor cells. When co-inoculated with tumor cells, macrophages with TFEB knockdown significantly enhanced tumor growth with increased infiltration of M2-like macrophages, reduced infiltration of CD8+ T cells, and enhanced angiogenesis in the tumors. Mechanistic studies revealed that TFEB downregulation resulted in macrophage M2 polarization through reducing SOCS3 production and enhancing STAT3 activation. We further demonstrate that the activation of TFEB by hydroxypropyl-ß-cyclodextrin in macrophages suppressed their M2 polarization and tumor-promoting capacity, and that macrophage-specific TFEB overexpression inhibited breast tumor growth in mice. Therefore, our data suggest that TFEB plays critical roles in macrophage polarization, and the downregulation of TFEB expression and activation is an integral part of tumor-induced immune editing in the TME. This study provides a rationale for a new cancer treatment strategy by modulating macrophage polarization through activating TFEB.

13.
Mol Cancer Ther ; 15(8): 1931-42, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27196773

RESUMEN

Macrophage infiltration correlates with severity in many types of cancer. Tumor cells recruit macrophages and educate them to adopt an M2-like phenotype through the secretion of chemokines and growth factors, such as MCP1 and CSF1. Macrophages in turn promote tumor growth through supporting angiogenesis, suppressing antitumor immunity, modulating extracellular matrix remodeling, and promoting tumor cell migration. Thus, tumor cells and macrophages interact to create a feedforward loop supporting tumor growth and metastasis. In this study, we tested the ability of emodin, a Chinese herb-derived compound, to inhibit breast cancer growth in mice and examined the underlying mechanisms. Emodin was used to treat mice bearing EO771 or 4T1 breast tumors. It was shown that emodin attenuated tumor growth by inhibiting macrophage infiltration and M2-like polarization, accompanied by increased T-cell activation and reduced angiogenesis in tumors. The tumor inhibitory effects of emodin were lost in tumor-bearing mice with macrophage depletion. Emodin inhibited IRF4, STAT6, and C/EBPß signaling and increased inhibitory histone H3 lysine 27 tri-methylation (H3K27m3) on the promoters of M2-related genes in tumor-associated macrophages. In addition, emodin inhibited tumor cell secretion of MCP1 and CSF1, as well as expression of surface anchoring molecule Thy-1, thus suppressing macrophage migration toward and adhesion to tumor cells. These results suggest that emodin acts on both breast cancer cells and macrophages and effectively blocks the tumor-promoting feedforward loop between the two cell types, thereby inhibiting breast cancer growth and metastasis. Mol Cancer Ther; 15(8); 1931-42. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/metabolismo , Comunicación Celular/efectos de los fármacos , Emodina/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Biomarcadores , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Ratones , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Oncoimmunology ; 5(11): e1232223, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27999745

RESUMEN

In antitumor immunity, dendritic cells (DCs) capture, process, and present tumor antigens to T cells, initiating a tumoricidal response. However, DCs are often dysfunctional due to their exposure to the tumor microenvironment (TME), leading to tumor escape from immune surveillance. Here, a vital role of microRNA-155 (miR-155) in regulating the function of DCs in breast cancer is reported. Host miR-155 deficiency enhanced breast cancer growth in mice, accompanied by reduced DCs in the tumors and draining lymph nodes. miR-155 deficiency in DCs impaired their maturation, migration ability, cytokine production, and the ability to activate T cells. We demonstrate that miR-155 regulates DC migration through epigenetic modulation of CCR7 expression. Moreover, IL-6 and IL-10, two cytokines abundant in the TME, are found to impair DC maturation by suppressing miR-155 expression. Furthermore, animal studies show that a lack of miR-155 diminishes the effectiveness of DC-based immunotherapy for breast cancer. In conclusion, these findings suggest that miR-155 is a master regulator of DC function in breast cancer, including maturation, cytokine secretion, migration toward lymph nodes, and activation of T-cells. These results suggest that boosting the expression of a single microRNA, miR-155, may significantly improve the efficacy of DC-based immunotherapies for breast cancer.

15.
Exp Biol Med (Maywood) ; 239(8): 1025-1035, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24740873

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is a major epidemics of the modern societies and has an inflammatory component in the pathogenesis. However, approved anti-inflammatory therapies are not currently available for the prevention of the transition from simple steatosis to non-alcoholic steatohepatitis (NASH). We aimed to test if a Chinese herb-derived compound, emodin could halt the simple steatosis to NASH transition. LDLR-/- mice were fed a western-type diet for 10 weeks; and during the last four weeks, the mice were intra-peritoneally injected daily with LPS with or without emodin. Systemic inflammation was evaluated by measurement of serum levels of cytokines and chemokines and flow cytometric analysis of spleen leukocytes. Liver inflammation was determined by histology, immunocytochemistry and flow cytometry. Quantitative real-time PCR and Western blot were performed to examine the effects of emodin on LPS-induced inflammatory responses in macrophages. Our data showed that emodin ameliorated systemic inflammation, reduced inflammatory cell infiltration in the liver, and attenuated liver function impairment. In vitro experiments showed emodin inhibited LPS-induced expression of proinflammatory cytokines in macrophages through suppressing Erk1/2 and p38 signaling. In conclusion, emodin inhibited the transition from simple steatosis to NASH in hyperlipidemic mice challenged with LPS through suppressing systemic and macrophage inflammation. Emodin may be developed as a therapy for NAFLD by the virtue of its anti-inflammatory effects.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda